Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Development ; 148(1)2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33318148

RESUMEN

Androgens/androgen receptor (AR)-mediated signaling pathways are essential for prostate development, morphogenesis and regeneration. Specifically, stromal AR signaling has been shown to be essential for prostatic initiation. However, the molecular mechanisms underlying AR-initiated mesenchymal-epithelial interactions in prostate development remain unclear. Here, using a newly generated mouse model, we have directly addressed the fate and role of genetically marked AR-expressing cells during embryonic prostate development. Androgen signaling-initiated signaling pathways were identified in mesenchymal niche populations at single-cell transcriptomic resolution. The dynamic cell-signaling networks regulated by stromal AR were additionally characterized in relation to prostatic epithelial bud formation. Pseudotime analyses further revealed the differentiation trajectory and fate of AR-expressing cells in both prostatic mesenchymal and epithelial cell populations. Specifically, the cellular properties of Zeb1-expressing progenitors were assessed. Selective deletion of AR signaling in a subpopulation of mesenchymal rather than epithelial cells dysregulated the expression of the master regulators and significantly impaired prostatic bud formation. These data provide novel, high-resolution evidence demonstrating the important role of mesenchymal androgen signaling in the cellular niche controlling prostate early development by initiating dynamic mesenchyme-epithelia cell interactions.


Asunto(s)
Andrógenos/farmacología , Comunicación Celular , Linaje de la Célula , Próstata/citología , Análisis de la Célula Individual , Animales , Comunicación Celular/efectos de los fármacos , Comunicación Celular/genética , Diferenciación Celular/efectos de los fármacos , Linaje de la Célula/efectos de los fármacos , Linaje de la Célula/genética , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Genes del Desarrollo , Masculino , Mesodermo/citología , Ratones , Próstata/efectos de los fármacos , RNA-Seq , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Células del Estroma/citología , Células del Estroma/efectos de los fármacos , Células del Estroma/metabolismo
2.
Development ; 148(19)2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34427305

RESUMEN

Stromal androgen-receptor (AR) action is essential for prostate development, morphogenesis and regeneration. However, mechanisms underlying how stromal AR maintains the cell niche in support of pubertal prostatic epithelial growth are unknown. Here, using advanced mouse genetic tools, we demonstrate that selective deletion of stromal AR expression in prepubescent Shh-responsive Gli1-expressing cells significantly impedes pubertal prostate epithelial growth and development. Single-cell transcriptomic analyses showed that AR loss in these prepubescent Gli1-expressing cells dysregulates androgen signaling-initiated stromal-epithelial paracrine interactions, leading to growth retardation of pubertal prostate epithelia and significant development defects. Specifically, AR loss elevates Shh-signaling activation in both prostatic stromal and adjacent epithelial cells, directly inhibiting prostatic epithelial growth. Single-cell trajectory analyses further identified aberrant differentiation fates of prostatic epithelial cells directly altered by stromal AR deletion. In vivo recombination of AR-deficient stromal Gli1-lineage cells with wild-type prostatic epithelial cells failed to develop normal prostatic epithelia. These data demonstrate previously unidentified mechanisms underlying how stromal AR-signaling facilitates Shh-mediated cell niches in pubertal prostatic epithelial growth and development.


Asunto(s)
Andrógenos/metabolismo , Proteínas Hedgehog/metabolismo , Próstata/crecimiento & desarrollo , Nicho de Células Madre , Animales , Diferenciación Celular , Linaje de la Célula , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/metabolismo , Proteínas Hedgehog/genética , Masculino , Ratones , Próstata/citología , Próstata/metabolismo , RNA-Seq , Receptores Androgénicos/genética , Receptores Androgénicos/metabolismo , Transducción de Señal , Análisis de la Célula Individual , Transcriptoma , Proteína con Dedos de Zinc GLI1/genética , Proteína con Dedos de Zinc GLI1/metabolismo
3.
Brief Bioinform ; 23(4)2022 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-35817303

RESUMEN

Many studies have proved that small nucleolar RNAs (snoRNAs) play critical roles in the development of various human complex diseases. Discovering the associations between snoRNAs and diseases is an important step toward understanding the pathogenesis and characteristics of diseases. However, uncovering associations via traditional experimental approaches is costly and time-consuming. This study proposed a bounded nuclear norm regularization-based method, called PSnoD, to predict snoRNA-disease associations. Benchmark experiments showed that compared with the state-of-the-art methods, PSnoD achieved a superior performance in the 5-fold stratified shuffle split. PSnoD produced a robust performance with an area under receiver-operating characteristic of 0.90 and an area under precision-recall of 0.55, highlighting the effectiveness of our proposed method. In addition, the computational efficiency of PSnoD was also demonstrated by comparison with other matrix completion techniques. More importantly, the case study further elucidated the ability of PSnoD to screen potential snoRNA-disease associations. The code of PSnoD has been uploaded to https://github.com/linDing-groups/PSnoD. Based on PSnoD, we established a web server that is freely accessed via http://psnod.lin-group.cn/.


Asunto(s)
Núcleo Celular , ARN Nucleolar Pequeño , Humanos , ARN Nucleolar Pequeño/genética
4.
Brief Bioinform ; 23(1)2022 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-34864888

RESUMEN

Post-translational modification (PTM) refers to the covalent and enzymatic modification of proteins after protein biosynthesis, which orchestrates a variety of biological processes. Detecting PTM sites in proteome scale is one of the key steps to in-depth understanding their regulation mechanisms. In this study, we presented an integrated method based on eXtreme Gradient Boosting (XGBoost), called iRice-MS, to identify 2-hydroxyisobutyrylation, crotonylation, malonylation, ubiquitination, succinylation and acetylation in rice. For each PTM-specific model, we adopted eight feature encoding schemes, including sequence-based features, physicochemical property-based features and spatial mapping information-based features. The optimal feature set was identified from each encoding, and their respective models were established. Extensive experimental results show that iRice-MS always display excellent performance on 5-fold cross-validation and independent dataset test. In addition, our novel approach provides the superiority to other existing tools in terms of AUC value. Based on the proposed model, a web server named iRice-MS was established and is freely accessible at http://lin-group.cn/server/iRice-MS.


Asunto(s)
Oryza , Procesamiento Proteico-Postraduccional , Acetilación , Biología Computacional , Modelos Biológicos , Oryza/metabolismo , Procesamiento Proteico-Postraduccional/fisiología , Proteoma/metabolismo , Ubiquitinación
5.
Brief Bioinform ; 22(5)2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-33751027

RESUMEN

DNase I hypersensitive site (DHS) refers to the hypersensitive region of chromatin for the DNase I enzyme. It is an important part of the noncoding region and contains a variety of regulatory elements, such as promoter, enhancer, and transcription factor-binding site, etc. Moreover, the related locus of disease (or trait) are usually enriched in the DHS regions. Therefore, the detection of DHS region is of great significance. In this study, we develop a deep learning-based algorithm to identify whether an unknown sequence region would be potential DHS. The proposed method showed high prediction performance on both training datasets and independent datasets in different cell types and developmental stages, demonstrating that the method has excellent superiority in the identification of DHSs. Furthermore, for the convenience of related wet-experimental researchers, the user-friendly web-server iDHS-Deep was established at http://lin-group.cn/server/iDHS-Deep/, by which users can easily distinguish DHS and non-DHS and obtain the corresponding developmental stage ofDHS.


Asunto(s)
Arabidopsis/genética , ADN/genética , Aprendizaje Profundo , Desoxirribonucleasa I/genética , Oryza/genética , Programas Informáticos , Arabidopsis/metabolismo , Cromatina/metabolismo , Cromatina/ultraestructura , ADN/química , ADN/metabolismo , Conjuntos de Datos como Asunto , Desoxirribonucleasa I/metabolismo , Elementos de Facilitación Genéticos , Sitios Genéticos , Humanos , Internet , Oryza/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética
6.
Curr Oncol Rep ; 25(9): 965-977, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37273124

RESUMEN

PURPOSE OF THE REVIEW: There have been increasing reports of cardiovascular complications of androgen deprivation therapy (ADT) leading to worse outcomes among patients with prostate cancer. While this may result from the direct effects of androgen suppression in the cardiovascular systems, there are ADT-type-specific distinct cardiovascular complications suggestive of mechanisms beyond androgen-mediated. Thus, it is critical to understand the biological and clinical impact of ADT on the cardiovascular system. RECENT FINDINGS: Gonadotropin-releasing hormone (GnRH) agonists cause increased cardiovascular events compared to GnRH antagonists. Androgen receptor antagonists are linked to an increased risk of long QT syndrome, torsades de pointes, and sudden cardiac death. Androgen synthesis inhibitors are associated with increased rates of hypertension, atrial tachyarrhythmia, and, in rare incidences, heart failure. ADT increases the risk of cardiovascular disease. The risk among ADT drugs differs and must be evaluated to develop a medically optimal plan for prostate cancer patients.


Asunto(s)
Sistema Cardiovascular , Neoplasias de la Próstata , Masculino , Humanos , Neoplasias de la Próstata/tratamiento farmacológico , Andrógenos/uso terapéutico , Antagonistas de Andrógenos/efectos adversos , Hormona Liberadora de Gonadotropina/agonistas , Hormona Liberadora de Gonadotropina/uso terapéutico , Biología
7.
Methods ; 203: 558-563, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34352373

RESUMEN

N4-methylcytosine (4mC) is a type of DNA modification which could regulate several biological progressions such as transcription regulation, replication and gene expressions. Precisely recognizing 4mC sites in genomic sequences can provide specific knowledge about their genetic roles. This study aimed to develop a deep learning-based model to predict 4mC sites in the Escherichia coli. In the model, DNA sequences were encoded by word embedding technique 'word2vec'. The obtained features were inputted into 1-D convolutional neural network (CNN) to discriminate 4mC sites from non-4mC sites in Escherichia coli genome. The examination on independent dataset showed that our model could yield the overall accuracy of 0.861, which was about 4.3% higher than the existing model. To provide convenience to scholars, we provided the data and source code of the model which can be freely download from https://github.com/linDing-groups/Deep-4mCW2V.


Asunto(s)
ADN , Escherichia coli , ADN/genética , Escherichia coli/genética , Genoma , Genómica , Programas Informáticos
8.
PLoS Genet ; 16(1): e1008588, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31929563

RESUMEN

Prostate embryonic development, pubertal and adult growth, maintenance, and regeneration are regulated through androgen signaling-mediated mesenchymal-epithelial interactions. Specifically, the essential role of mesenchymal androgen signaling in the development of prostate epithelium has been observed for over 30 years. However, the identity of the mesenchymal cells responsible for this paracrine regulation and related mechanisms are still unknown. Here, we provide the first demonstration of an indispensable role of the androgen receptor (AR) in sonic hedgehog (SHH) responsive Gli1-expressing cells, in regulating prostate development, growth, and regeneration. Selective deletion of AR expression in Gli1-expressing cells during embryogenesis disrupts prostatic budding and impairs prostate development and formation. Tissue recombination assays showed that urogenital mesenchyme (UGM) containing AR-deficient mesenchymal Gli1-expressing cells combined with wildtype urogenital epithelium (UGE) failed to develop normal prostate tissue in the presence of androgens, revealing the decisive role of AR in mesenchymal SHH responsive cells in prostate development. Prepubescent deletion of AR expression in Gli1-expressing cells resulted in severe impairment of androgen-induced prostate growth and regeneration. RNA-sequencing analysis showed significant alterations in signaling pathways related to prostate development, stem cells, and organ morphogenesis in AR-deficient Gli1-expressing cells. Among these altered pathways, the transforming growth factor ß1 (TGFß1) pathway was up-regulated in AR-deficient Gli1-expressing cells. We further demonstrated the activation of TGFß1 signaling in AR-deleted prostatic Gli1-expressing cells, which inhibits prostate epithelium growth through paracrine regulation. These data demonstrate a novel role of the AR in the Gli1-expressing cellular niche for regulating prostatic cell fate, morphogenesis, and renewal, and elucidate the mechanism by which mesenchymal androgen-signaling through SHH-responsive cells elicits the growth and regeneration of prostate epithelium.


Asunto(s)
Proteínas Hedgehog/metabolismo , Morfogénesis , Próstata/metabolismo , Receptores Androgénicos/metabolismo , Regeneración , Transducción de Señal , Animales , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/metabolismo , Masculino , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Ratones , Próstata/citología , Próstata/crecimiento & desarrollo , Próstata/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Proteína con Dedos de Zinc GLI1/genética , Proteína con Dedos de Zinc GLI1/metabolismo
9.
PLoS Genet ; 15(10): e1008451, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31658259

RESUMEN

E-cadherin complexes with the actin cytoskeleton via cytoplasmic catenins and maintains the functional characteristics and integrity of the epithelia in normal epithelial tissues. Lost expression of E-cadherin disrupts this complex resulting in loss of cell polarity, epithelial denudation and increased epithelial permeability in a variety of tissues. Decreased expression of E-cadherin has also been observed in invasive and metastatic human tumors. In this study, we investigated the effect of E-cadherin loss in prostatic epithelium using newly developed genetically engineered mouse models. Deletion of E-cadherin in prostatic luminal epithelial cells with modified probasin promoter driven Cre (PB-Cre4) induced the development of mouse prostatic intraepithelial neoplasia (PIN). An increase in levels of cytoplasmic and nuclear ß-catenin appeared in E-cadherin deleted atypical cells within PIN lesions. Using various experimental approaches, we further demonstrated that the knockdown of E-cadherin expression elevated free cytoplasmic and nuclear ß-catenin and enhanced androgen-induced transcription and cell growth. Intriguingly, pathological changes representing prostatic epithelial cell denudation and increased apoptosis accompanied the above PIN lesions. The essential role of E-cadherin in maintaining prostatic epithelial integrity and organization was further demonstrated using organoid culture approaches. To directly assess the role of loss of E-cadherin in prostate tumor progression, we generated a new mouse model with bigenic Cdh1 and Pten deletion in prostate epithelium. Early onset, aggressive tumor phenotypes presented in the compound mice. Strikingly, goblet cell metaplasia was observed, intermixed within prostatic tumor lesions of the compound mice. This study provides multiple lines of novel evidence demonstrating a comprehensive role of E-cadherin in maintaining epithelial integrity during the course of prostate oncogenic transformation, tumor initiation and progression.


Asunto(s)
Antígenos CD/metabolismo , Cadherinas/metabolismo , Transformación Celular Neoplásica/patología , Neoplasia Intraepitelial Prostática/patología , Neoplasias de la Próstata/patología , Animales , Antígenos CD/genética , Cadherinas/genética , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Epiteliales , Epitelio , Células HEK293 , Humanos , Masculino , Ratones , Ratones Transgénicos , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Cultivo Primario de Células , Próstata/citología , Próstata/patología , Neoplasia Intraepitelial Prostática/genética , Neoplasias de la Próstata/genética , ARN Interferente Pequeño , beta Catenina/genética , beta Catenina/metabolismo
10.
Ecotoxicol Environ Saf ; 241: 113731, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35688001

RESUMEN

Triphenyl phosphate (TPhP) is used as a flame retardant that gradually leaks from products into the marine environment and thus may threaten low-trophic-level marine organisms, such as zooplankton. To assess the effect of TPhP on these taxa, we treated the marine rotifer Brachionus plicatilis as a target and examined the changes in key life history parameters and the metabolome after exposure to TPhP at 0.02, 1 and 5 mg/L. Additionally, the rotifer-Phaeocystis population dynamics (a simulation of the prey-predator relationship) were studied under TPhP stress. Our results showed that TPhP at 1 and 5 mg/L reduced the average lifespan and the total offspring number and prolonged the prereproductive time, suggesting damage to survival and fecundity. In the 0.02 mg/L group, no obvious damage occurred in the overall condition of rotifers, but the volume of parental rotifers after the first brood decreased. This implied that rotifers sacrificed somatic growth to reproduction in the initial period of TPhP exposure at the low concentration. All the tested TPhP concentrations altered the rotifer-Phaeocystis population dynamic changes, especially that 1 mg/L TPhP reduced the ability of rotifers to remove this harmful alga, as evidenced by the decrease in the maximum population density of rotifers and the extended time to P. globosa extinction. At the molecular level, metabolomics identified 84 and 206 differentially expressed metabolites, most of which were enriched in glycerophospholipid metabolism, steroid biosynthesis and sphingolipid metabolism. Nile red staining showed a decrease in neutral lipids in rotifers, further indicating a disorder of lipid metabolism induced by TPhP. Moreover, the balance between ROS production and the defense system was disrupted by TPhP, which contributed to its toxicity. This finding will promote the understanding of the ecological risk and mode of action of TPhP in aquatic environments.


Asunto(s)
Haptophyta , Rotíferos , Animales , Organofosfatos/metabolismo , Dinámica Poblacional
11.
Int J Mol Sci ; 23(9)2022 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-35563328

RESUMEN

As emerging pollutants, microplastics (MPs) and organophosphorus esters (OPEs) coexist in the aquatic environment, posing a potential threat to organisms. Although toxicological studies have been conducted individually, the effects of combined exposure are unknown since MPs can interact with OPEs acting as carriers. In this study, we assessed the response of marine rotifer, Brachionus plicatilis, to co-exposure to polystyrene MPs and tris(2-chloroethyl) phosphate (TCEP) at different concentrations, including population growth, oxidative status, and transcriptomics. Results indicated that 0.1 µm and 1 µm MPs were accumulated in the digestive system, and, even at up to 2000 µg/L, they did not exert obvious damage to the stomach morphology, survival, and reproduction of B. plicatilis. The presence of 1 µm MPs reversed the low population growth rate and high oxidative stress induced by TCEP to the normal level. Some genes involved in metabolic detoxification and stress response were upregulated, such as ABC and Hsp. Subsequent validation showed that P-glycoprotein efflux ability was activated by combined exposure, indicating its important role in the reversal of population growth inhibition. Such results challenge the common perception that MPs aggravate the toxicity of coexisting pollutants and elucidate the molecular mechanism of the limited toxic effects induced by MPs and TCEP.


Asunto(s)
Rotíferos , Contaminantes Químicos del Agua , Animales , Microplásticos/toxicidad , Fosfatos/metabolismo , Plásticos/metabolismo , Poliestirenos/farmacología , Contaminantes Químicos del Agua/metabolismo
12.
Int J Mol Sci ; 23(3)2022 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-35163174

RESUMEN

4mC is a type of DNA alteration that has the ability to synchronize multiple biological movements, for example, DNA replication, gene expressions, and transcriptional regulations. Accurate prediction of 4mC sites can provide exact information to their hereditary functions. The purpose of this study was to establish a robust deep learning model to recognize 4mC sites in Geobacter pickeringii. In the anticipated model, two kinds of feature descriptors, namely, binary and k-mer composition were used to encode the DNA sequences of Geobacter pickeringii. The obtained features from their fusion were optimized by using correlation and gradient-boosting decision tree (GBDT)-based algorithm with incremental feature selection (IFS) method. Then, these optimized features were inserted into 1D convolutional neural network (CNN) to classify 4mC sites from non-4mC sites in Geobacter pickeringii. The performance of the anticipated model on independent data exhibited an accuracy of 0.868, which was 4.2% higher than the existing model.


Asunto(s)
Biología Computacional/métodos , Epigénesis Genética/genética , Geobacter/genética , Algoritmos , Citosina/metabolismo , ADN/genética , Metilación de ADN/genética , Aprendizaje Profundo , Aprendizaje Automático , Mutación/genética , Redes Neurales de la Computación , Programas Informáticos
13.
J Biol Chem ; 295(2): 631-644, 2020 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-31819003

RESUMEN

Co-occurrence of aberrant hepatocyte growth factor (HGF)/MET proto-oncogene receptor tyrosine kinase (MET) and Wnt/ß-catenin signaling pathways has been observed in advanced and metastatic prostate cancers. This co-occurrence positively correlates with prostate cancer progression and castration-resistant prostate cancer development. However, the biological consequences of these abnormalities in these disease processes remain largely unknown. Here, we investigated the aberrant activation of HGF/MET and Wnt/ß-catenin cascades in prostate tumorigenesis by using a newly generated mouse model in which both murine Met transgene and stabilized ß-catenin are conditionally co-expressed in prostatic epithelial cells. These compound mice displayed accelerated prostate tumor formation and invasion compared with their littermates that expressed only stabilized ß-catenin. RNA-Seq and quantitative RT-PCR analyses revealed increased expression of genes associated with tumor cell proliferation, progression, and metastasis. Moreover, Wnt signaling pathways were robustly enriched in prostate tumor samples from the compound mice. ChIP-qPCR experiments revealed increased ß-catenin recruitment within the regulatory regions of the Myc gene in tumor cells of the compound mice. Interestingly, the occupancy of MET on the Myc promoter also appeared in the compound mouse tumor samples, implicating a novel role of MET in ß-catenin-mediated transcription. Results from implanting prostate graft tissues derived from the compound mice and controls into HGF-transgenic mice further uncovered that HGF induces prostatic oncogenic transformation and cell growth. These results indicate a role of HGF/MET in ß-catenin-mediated prostate cancer cell growth and progression and implicate a molecular mechanism whereby nuclear MET promotes aberrant Wnt/ß-catenin signaling-mediated prostate tumorigenesis.


Asunto(s)
Factor de Crecimiento de Hepatocito/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal , beta Catenina/metabolismo , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Humanos , Masculino , Ratones Endogámicos C57BL , Ratones SCID , Próstata/metabolismo , Próstata/patología , Neoplasias de la Próstata/patología , Proto-Oncogenes Mas
14.
Differentiation ; 107: 1-10, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30927641

RESUMEN

Androgen signaling is essential for prostate development, morphogenesis, and regeneration. Emerging evidence also indicates a regulatory role of Notch signaling in prostate development, differentiation, and growth. However, the collaborative regulatory mechanisms of androgen and Notch signaling during prostate development, growth, and regeneration are largely unknown. Hairy and Enhancer of Split 1 (Hes1) is a transcriptional regulator of Notch signaling pathways, and its expression is responsive to Notch signaling. Hes1-expressing cells have been shown to possess the regenerative capability to repopulate a variety of adult tissues. In this study, we developed new mouse models to directly assess the role of the androgen receptor in prostatic Hes1-expressing cells. Selective deletion of AR expression in embryonic Hes1-expressing cells impeded early prostate development both in vivo and in tissue xenograft experiments. Prepubescent deletion of AR expression in Hes1-expressing cells resulted in prostate glands containing abnormalities in cell morphology and gland architecture. A population of castration-resistant Hes1-expressing cells was revealed in the adult prostate, with the ability to repopulate prostate epithelium following androgen supplementation. Deletion of AR in Hes1-expressing cells diminishes their regenerative ability. These lines of evidence demonstrate a critical role for the AR in Notch-responsive cells during the course of prostate development, morphogenesis, and regeneration, and implicate a mechanism underlying interaction between the androgen and Notch signaling pathways in the mouse prostate.


Asunto(s)
Próstata/fisiología , Receptores Notch/metabolismo , Regeneración , Factor de Transcripción HES-1 , Andrógenos/metabolismo , Animales , Masculino , Ratones , Modelos Animales , Próstata/embriología , Receptores Androgénicos/metabolismo , Transducción de Señal , Factor de Transcripción HES-1/biosíntesis , Factor de Transcripción HES-1/genética
15.
J Biol Chem ; 293(52): 20123-20136, 2018 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-30401749

RESUMEN

Emerging evidence has shown that the hepatocyte growth factor (HGF) and its receptor, MET proto-oncogene, receptor tyrosine kinase (MET), promote cell proliferation, motility, morphogenesis, and angiogenesis. Whereas up-regulation of MET expression has been observed in aggressive and metastatic prostate cancer, a clear understanding of MET function in prostate tumorigenesis remains elusive. Here, we developed a conditional Met transgenic mouse strain, H11Met/+:PB-Cre4, to mimic human prostate cancer cells with increased MET expression in the prostatic luminal epithelium. We found that these mice develop prostatic intraepithelial neoplasia after HGF administration. To further assess the biological role of MET in prostate cancer progression, we bred H11Met/+/PtenLoxP/LoxP:PBCre4 compound mice, in which transgenic Met expression and deletion of the tumor suppressor gene Pten occurred simultaneously only in prostatic epithelial cells. These compound mice exhibited accelerated prostate tumor formation and invasion as well as increased metastasis compared with PtenLoxP/LoxP:PB-Cre4 mice. Moreover, prostatic sarcomatoid carcinomas and lesions resembling the epithelial-to-mesenchymal transition developed in tumor lesions of the compound mice. RNA-Seq and qRT-PCR analyses revealed a robust enrichment of known tumor progression and metastasis-promoting genes in samples isolated from H11Met/+/PtenLoxP/LoxP:PB-Cre4 compound mice compared with those from PtenLoxP/LoxP:PB-Cre4 littermate controls. HGF-induced cell proliferation and migration also increased in mouse embryonic fibroblasts (MEFs) from animals with both Met transgene expression and Pten deletion compared with Pten-null MEFs. The results from these newly developed mouse models indicate a role for MET in hastening tumorigenesis and metastasis when combined with the loss of tumor suppressors.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Neoplasias de la Próstata/metabolismo , Proteínas Proto-Oncogénicas c-met/metabolismo , Transducción de Señal , Animales , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Factor de Crecimiento de Hepatocito/genética , Masculino , Ratones , Ratones Transgénicos , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas c-met/genética
16.
Stem Cells ; 36(6): 891-902, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29451339

RESUMEN

Androgen signaling is essential for prostate development, morphogenesis, and regeneration. Emerging evidence indicates that Wnt/ß-catenin signaling also contributes to prostate development specifically through regulation of cell fate determination. Prostatic Axin2-expressing cells are able to respond to Wnt signals and possess the progenitor properties to regenerate prostatic epithelium. Despite critical roles of both signaling pathways, the biological significance of androgen receptor (AR) in Axin2-expressing/Wnt-responsive cells remains largely unexplored. In this study, we investigated this important question using a series of newly generated mouse models. Deletion of Ar in embryonic Axin2-expressing cells impaired early prostate development in both ex vivo and tissue implantation experiments. When Ar expression was deleted in prostatic Axin2-expressing cells at pre-puberty stages, it results in smaller and underdeveloped prostates. A subpopulation of Axin2 expressing cells in prostate epithelium is resistant to castration and, following androgen supplementation, is capable to expand to prostatic luminal cells. Deletion of Ar in these Axin2-expressing cells reduces their regenerative ability. These lines of evidence demonstrate an indispensable role for the Ar in Wnt-responsive cells during the course of prostate development, morphogenesis, and regeneration, which also imply an underlying interaction between the androgen and Wnt signaling pathways in the mouse prostate. Stem Cells 2018;36:891-902.


Asunto(s)
Próstata/fisiología , Receptores Androgénicos/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Proliferación Celular , Humanos , Masculino , Morfogénesis , Regeneración
17.
J Biol Chem ; 290(5): 2759-68, 2015 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-25527506

RESUMEN

Both androgen action and PI3K medicated signaling pathways have been implicated in prostate tumorigenesis. Our androgen receptor (AR) conditional transgenic mice developed murine prostatic intraepithelial neoplasia (mPIN) and prostatic adenocarcinoma lesions recapitulating human prostate cancer development and progression. Role of transgenic AR contributing to malignancy was demonstrated by high degree of transgenic AR expression in atypical and tumor cells in mPIN as well as prostatic adenocarcinoma lesions of the transgenic mice, but not in adjacent normal tissue. Interestingly, reduced PI3K/Akt activation also appeared in these mouse atypical and tumor cells, suggesting an interaction between androgen and PI3K/AKT pathways. In this study, we further investigated this interaction. We showed that the androgen depletion or knockdown of AR expression results in elevated levels of active phosphorylated AKT in prostate cancer cells. Castration of conditional Pten knock-out mice showed increased Akt, phosphorylated Akt, and pS6 expression in the mouse prostate. Using a series of newly generated Ar reporter and Pten knock-out compound mice, we showed that Pten loss directly represses endogenous Ar expression in prostatic epithelial cells. Moreover, Pten loss and PI3K/Akt activation reduced Ar-mediated transcription in purified Pten-null cells. This study provides novel evidence demonstrating interplay between androgen and PI3K pathways, as well as introduces unique and relevant mouse models for further studies of PI3K and AR pathways in the context of prostate tumorigenesis.


Asunto(s)
Fosfatidilinositol 3-Quinasas/metabolismo , Próstata/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Andrógenos/metabolismo , Animales , Western Blotting , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Noqueados , Neoplasias de la Próstata/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/fisiología
18.
Stem Cells ; 33(11): 3356-67, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26220362

RESUMEN

The precise role of Wnt/ß-catenin signaling during prostatic development and tumorigenesis is unclear. Axin2 is a direct transcriptional target of ß-catenin. Recent studies have shown that Axin2-expressing cells have stem/progenitor cell properties in a variety of mouse tissues. Here, we genetically labeled Axin2-expressing cells at various time points and tracked their cellular behavior at different developmental and mature stages. We found that prostatic Axin2-expressing cells mainly express luminal epithelial cell markers and are able to expand luminal cell lineages during prostatic development and maturation. They can also survive androgen withdrawal and regenerate prostatic luminal epithelial cells following androgen replacement. Deletion of ß-catenin or expression of stabilized ß-catenin in these Axin2-expressing cells results in abnormal development or oncogenic transformation, respectively. Our study uncovers a critical role of Wnt/ß-catenin-responsive cells in prostatic development and regeneration, and that dysregulation of Wnt/ß-catenin signaling in these cells contributes to prostatic developmental defects and tumorigenesis.


Asunto(s)
Proteína Axina/biosíntesis , Próstata/crecimiento & desarrollo , Próstata/metabolismo , Regeneración/fisiología , Vía de Señalización Wnt/fisiología , beta Catenina/biosíntesis , Animales , Linaje de la Célula , Células Epiteliales/metabolismo , Masculino , Ratones , Ratones Transgénicos , Organogénesis/fisiología , Próstata/citología
19.
J Biol Chem ; 288(50): 35913-24, 2013 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-24174533

RESUMEN

ZMIZ2, also named ZIMP7, is a protein inhibitor of activated STAT (PIAS)-like protein and a transcriptional coactivator. In this study, we investigated the interaction between ZMIZ2 and ß-catenin, a key regulator of the Wnt signaling pathway. We demonstrated that the expression of exogenous ZMIZ2 augments TCF (T cell factor) and ß-catenin-mediated transcription. In contrast, shRNA knockdown of ZMIZ2 expression specifically represses the enhancement of TCF/ß-catenin-mediated transcription by ZMIZ2. Using Wnt3a-conditioned medium, we demonstrated that ZMIZ2 can enhance Wnt ligand-induced TCF/ß-catenin-mediated transcription. We also showed a promotional role of ZMIZ2 in enhancing ß-catenin downstream target gene expression in human cells and in Zmiz2 null (Zmiz2(-/-)) mouse embryonic fibroblasts (MEFs). The regulatory role of Zmiz2 in Wnt-induced TCF/ß-catenin-mediated transcription can be restored in Zmiz2(-/-) MEFs that were infected with adenoviral expression vectors for Zmiz2. Moreover, enhancement of Zmiz2 on TCF/ß-catenin-mediated transcription was further demonstrated in Zmiz2 knockout and Axin2 reporter compound mice. Furthermore, the protein-protein interaction between ZMIZ2 and ß-catenin was identified by co-immunoprecipitation and in vitro protein pulldown assays. We also observed recruitment of endogenous ZMIZ2 onto the promoter region of the Axin 2 gene, a ß-catenin downstream target promoter, in a Wnt ligand-inducible manner. Finally, a promotional role of ZMIZ2 on cell growth was demonstrated in human cell lines and Zmiz2 knockout MEFs. Our findings demonstrate a novel interaction between ZMIZ2 and ß-catenin and elucidate a novel mechanism for PIAS-like proteins in regulating Wnt signaling pathways.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Proteína Axina/genética , Línea Celular , Proliferación Celular , Femenino , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Receptores de Hialuranos/metabolismo , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Regiones Promotoras Genéticas/genética , Proteínas Inhibidoras de STAT Activados , Factores de Transcripción TCF/metabolismo , Transcripción Genética
20.
J Biol Chem ; 288(6): 3727-38, 2013 Feb 08.
Artículo en Inglés | MEDLINE | ID: mdl-23275340

RESUMEN

Using an Lzts2 knock-out mouse model, we characterized the biological role of Lzts2 in tumorigenesis. Both heterozygous and homozygous deletion of the Lzts2-targeted allele in mice shows an increased incidence in spontaneous tumor development, although Lzts2 homozygous knock-out mice show significantly higher incidences than heterozygous mice. Treatment of Lzts2-deficient mice with a carcinogen, N-butyl-N-(4-hydroxybutyl) nitrosamine, increases the susceptibility to N-butyl-N-(4-hydroxybutyl) nitrosamine-induced bladder carcinoma development. Examination of human prostate cancer tissue specimens shows a reduction of LZTS2 protein expression in prostate cancer cells. Further analyses of mouse embryonic fibroblasts isolated from Lzts2 knock-out embryos show that loss of Lzts2 enhances cell growth. These data provide the first line of evidence demonstrating that deletion of Lzts2 increases susceptibility to spontaneous and carcinogen-induced tumor development.


Asunto(s)
Proteínas de Ciclo Celular/biosíntesis , Proteínas de Unión al ADN/biosíntesis , Eliminación de Gen , Predisposición Genética a la Enfermedad , Neoplasias de la Próstata/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Neoplasias de la Vejiga Urinaria/metabolismo , Animales , Butilhidroxibutilnitrosamina/toxicidad , Carcinógenos/toxicidad , Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Células HEK293 , Heterocigoto , Humanos , Masculino , Ratones , Ratones Noqueados , Neoplasias de la Próstata/inducido químicamente , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Proteínas Supresoras de Tumor/genética , Neoplasias de la Vejiga Urinaria/inducido químicamente , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA